1
|
Piki E, Dini A, Rantanen F, Bentz F, Paavolainen L, Barker H, Raivola J, Hirasawa A, Kallioniemi O, Murumägi A, Ungureanu D. Molecular and functional profiling of primary normal ovarian cells defines insights into cancer development and drug responses. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200903. [PMID: 39634630 PMCID: PMC11616607 DOI: 10.1016/j.omton.2024.200903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 10/11/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024]
Abstract
Patients with ovarian cancer, especially the high-grade serous ovarian cancer (HGSOC) subtype, face poor prognosis due to late diagnosis and treatment resistance. Owing to the high heterogeneity of HGSOC, identifying the origin of the disease and optimal treatments is difficult. Here, we characterized two primary immortalized human ovarian cell lines, human ovarian surface epithelium (HOSE)1C and HOSE2C, comparing their molecular profiling with representative HGSOC cells. We identified molecular features associated with normal and malignant phenotype of ovarian cells by applying single-cell transcriptomics, high-content image-based cell painting, and high-throughput drug testing. Our findings reveal distinct transcriptomic and morphological profiles for the two HOSEs, with a stromal phenotype. Moreover, their responses to the tumor microenvironment differ, exemplified by STAT1 and GREM1 upregulation in HOSE1C and HOSE2C, respectively. We identified selective activation of ERK/MEK targeted inhibitors in cancer cells compared to HOSEs. This study offers insights into the normal and malignant ovarian cells, shedding light on cancer development and drug responses.
Collapse
Affiliation(s)
- Emilia Piki
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Alice Dini
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Frida Rantanen
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Franziska Bentz
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00014 Helsinki, Finland
| | - Lassi Paavolainen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00014 Helsinki, Finland
| | - Harlan Barker
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
- Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| | - Juuli Raivola
- Applied Tumor Genomics, Research Program Unit, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Akira Hirasawa
- Department of Clinical Genomic Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00014 Helsinki, Finland
- Science for Life Laboratory (SciLifeLab), Department of Oncology and Pathology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Astrid Murumägi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00014 Helsinki, Finland
| | - Daniela Ungureanu
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
2
|
Bolatimi OE, Hua Y, Ekuban FA, Gripshover TC, Ekuban A, Luulay B, Watson WH, Hardesty JE, Wahlang B. Low dose exposure to dioxins alters hepatic energy metabolism and steatotic liver disease development in a sex-specific manner. ENVIRONMENT INTERNATIONAL 2024; 194:109152. [PMID: 39577358 PMCID: PMC11700233 DOI: 10.1016/j.envint.2024.109152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/09/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024]
Abstract
"Dioxins" are persistent organic pollutants (POPs) that are continuously present in the environment at appreciable levels and have been associated with increased risk of steatotic liver disease (SLD). However, current understanding of the role of sex and effects of mixtures of dioxins in SLD development is limited. Additionally, there exists debates on the levels of dioxins required to be considered dangerous as emphasis has shifted from high level exposure events to the steady state of lower-level exposures. We therefore investigated sex-dependent effects of low-level exposures to a mixture of dioxins: 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), 2,3,4,7,8-Pentachlorodibenzofuran (PeCDF) and Polychlorinated biphenyl 126 (PCB126), in the context of SLD and associated metabolic dysfunction. Male and female C57BL/6J mice were fed a low-fat diet and weekly administered either vehicle control or TCDD (10 ng/kg), PeCDF (80 ng/kg) and PCB 126 (140 ng/kg) over a two-week period. Female mice generally demonstrated higher hepatic fat content compared to males. However, exposure to dioxins further elevated hepatic cholesterol levels in females, and this was accompanied by increased lipogenic gene expression (Acaca, Fasn) in the liver. In contrast, exposed males but not females displayed higher white adipose tissue weights. Furthermore, TCDD + PeCDF + PCB126 activated the AHR (hepatic Cyp1a1, Cyp1a2 induction); with Cyp1a1 induction observed only in exposed females. Notably, gene expression of hepatic albumin (Alb) was also reduced only in exposed females. Overall, exposure to the low dose dioxin mixture compromised hepatic homeostasis via metabolic perturbations, and hepatic dysregulation was more accelerated in female livers.
Collapse
Affiliation(s)
- Oluwanifemi E Bolatimi
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA; University of Louisville (UofL) Superfund Research Center, University of Louisville, Louisville, KY 40202, USA
| | - Yuan Hua
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Frederick A Ekuban
- University of Louisville (UofL) Superfund Research Center, University of Louisville, Louisville, KY 40202, USA; Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Tyler C Gripshover
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA; Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Abigail Ekuban
- University of Louisville (UofL) Superfund Research Center, University of Louisville, Louisville, KY 40202, USA; Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Bana Luulay
- College of Arts and Sciences, University of Louisville, Louisville, KY 40202, USA
| | - Walter H Watson
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA; Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, KY 40202, USA; The Hepatobiology & Toxicology Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; Alcohol Research Center, University of Louisville, Louisville, KY 40202, USA; The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, KY 40202, USA
| | - Josiah E Hardesty
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA; Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, KY 40202, USA; The Hepatobiology & Toxicology Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; Alcohol Research Center, University of Louisville, Louisville, KY 40202, USA
| | - Banrida Wahlang
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA; University of Louisville (UofL) Superfund Research Center, University of Louisville, Louisville, KY 40202, USA; Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, KY 40202, USA; The Hepatobiology & Toxicology Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
3
|
Gan G, Zhang R, Zeng Y, Lu B, Luo Y, Chen S, Lei H, Cai Z, Huang X. Fecal microbiota transplantation validates the importance of gut microbiota in an ApoE -/- mouse model of chronic apical periodontitis-induced atherosclerosis. BMC Oral Health 2024; 24:1455. [PMID: 39614243 DOI: 10.1186/s12903-024-05230-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Chronic apical periodontitis (CAP) has been linked to the development of atherosclerosis, although the underlying mechanisms remain unclear. This study aimed to investigate the role of gut microbiota disruption in CAP-induced atherosclerosis development, focusing on trimethylamine N-oxide (TMAO)-related metabolites. METHODS The study utilized fecal microbiota transplantation (FMT) to transfer gut microbiota from mice with CAP to healthy mice. Atherosclerosis development was assessed by analyzing lesions in the aortic arch and aortic root. Serum lipid and inflammatory factor levels were measured. Composition and diversity of gut microbiota were analyzed using targeted metabolomics, with a focus on the ratio of Firmicutes to Bacteroidetes. The expression of hepatic flavin-containing monooxygenase 3 (FMO3) and serum TMAO levels were also evaluated. RESULTS Mice receiving gut microbiota from CAP mice showed increased atherosclerotic lesions compared to controls, without significant differences in serum lipid or inflammatory factor levels. Alterations in gut microbiota composition were observed, characterized by an increase in the Firmicutes to Bacteroidetes ratio. Peptostreptococcaceae abundance positively correlated with atherosclerosis severity, while Odoribacteraceae showed a negative correlation. No significant differences were found in hepatic FMO3 expression or serum TMAO levels. CONCLUSIONS The study confirms the role of gut microbiota disruption in CAP-mediated atherosclerosis development, independent of serum lipid or TMAO levels. Alterations in gut microbiota composition, particularly increased Firmicutes to Bacteroidetes ratio and specific bacterial families, were associated with atherosclerosis severity. These findings highlight the intricate interplay between gut microbiota and cardiovascular health in the context of CAP.
Collapse
Affiliation(s)
- Guowu Gan
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Ren Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yu Zeng
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Beibei Lu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yufang Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shuai Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Huaxiang Lei
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Zhiyu Cai
- Department of Stomatology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaojing Huang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
4
|
Sink WJ, Fling R, Yilmaz A, Nault R, Goniwiecha D, Harkema JR, Graham SF, Zacharewski T. 2,3,7,8-Tetrachlorodibenzo- p-dioxin (TCDD) elicited dose-dependent shifts in the murine urinary metabolome associated with hepatic AHR-mediated differential gene expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619714. [PMID: 39484576 PMCID: PMC11526911 DOI: 10.1101/2024.10.22.619714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Epidemiological evidence suggests an association between dioxin and dioxin-like compound (DLC) exposure and human liver disease. The prototypical DLC, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), has been shown to induce the progression of reversible hepatic steatosis to steatohepatitis with periportal fibrosis and biliary hyperplasia in mice. Although the effects of TCDD toxicity are mediated by aryl hydrocarbon receptor (AHR) activation, the underlying mechanisms of TCDD-induced hepatotoxicity are unresolved. In the present study, male C57BL/6NCrl mice were gavaged every 4 days for 28 days with 0.03 - 30 μg/kg TCDD and evaluated for liver histopathology and gene expression as well as complementary 1-dimensional proton magnetic resonance (1D- 1H NMR) urinary metabolic profiling. Urinary trimethylamine (TMA), trimethylamine N-oxide (TMAO), and 1-methylnicotinamide (1MN) levels were altered by TCDD at doses ≤ 3 μg/kg; other urinary metabolites, like glycolate, urocanate, and 3-hydroxyisovalerate, were only altered at doses that induced moderate to severe steatohepatitis. Bulk liver RNA-seq data suggested altered urinary metabolites correlated with hepatic differential gene expression corresponding to specific metabolic pathways. In addition to evaluating whether altered urinary metabolites were liver-dependent, published single-nuclear RNA-seq (snRNA-seq), AHR ChIP-seq, and AHR knockout gene expression datasets provide further support for hepatic cell-type and AHR-regulated dependency, respectively. Overall, TCDD-induced liver effects were preceded by and occurred with changes in urinary metabolite levels due to AHR-mediated changes in hepatic gene expression.
Collapse
Affiliation(s)
- Warren J Sink
- Michigan State University, Department of Biochemistry and Molecular Biology, East Lansing, MI 48823, USA
- Michigan State University, Institute for Integrative Toxicology, East Lansing, MI 48824, USA
| | - Russell Fling
- Michigan State University, Department of Biochemistry and Molecular Biology, East Lansing, MI 48823, USA
- Michigan State University, Institute for Integrative Toxicology, East Lansing, MI 48824, USA
| | - Ali Yilmaz
- Corewell Health Research Institute, Royal Oak, MI 48073, USA
| | - Rance Nault
- Michigan State University, Department of Pharmacology and Toxicology, East Lansing, MI 48824, USA
| | - Delanie Goniwiecha
- Middlebury College, Neuroscience Faculty, 14 Old Chapel Rd, Middlebury, VT 05753, USA
| | - Jack R Harkema
- Michigan State University, Pathobiology & Diagnostic Investigation, East Lansing, MI, United States of America
| | - Stewart F Graham
- Corewell Health Research Institute, Royal Oak, MI 48073, USA
- Oakland University-William Beaumont School of Medicine, Rochester, MI 48309, USA
| | - Timothy Zacharewski
- Michigan State University, Department of Biochemistry and Molecular Biology, East Lansing, MI 48823, USA
- Michigan State University, Institute for Integrative Toxicology, East Lansing, MI 48824, USA
| |
Collapse
|
5
|
Cholico GN, Nault R, Zacharewski T. Cell-specific AHR-driven differential gene expression in the mouse liver cell following acute TCDD exposure. BMC Genomics 2024; 25:809. [PMID: 39198768 PMCID: PMC11351262 DOI: 10.1186/s12864-024-10730-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024] Open
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a persistent environmental contaminant that disrupts hepatic function leading to steatotic liver disease (SLD)-like pathologies, such as steatosis, steatohepatitis, and fibrosis. These effects are mediated by the aryl hydrocarbon receptor following changes in gene expression. Although diverse cell types are involved, initial cell-specific changes in gene expression have not been reported. In this study, differential gene expression in hepatic cell types was examined in male C57BL/6 mice gavaged with 30 µg/kg of TCDD using single-nuclei RNA-sequencing. Ten liver cell types were identified with the proportions of most cell types remaining unchanged, except for neutrophils which increased at 72 h. Gene expression suggests TCDD induced genes related to oxidative stress in hepatocytes as early as 2 h. Lipid homeostasis was disrupted in hepatocytes, macrophages, B cells, and T cells, characterized by the induction of genes associated with lipid transport, steroid hormone biosynthesis, and the suppression of β-oxidation, while linoleic acid metabolism was altered in hepatic stellate cells (HSCs), B cells, portal fibroblasts, and plasmacytoid dendritic cells. Pro-fibrogenic processes were also enriched, including the induction retinol metabolism genes in HSCs and the early induction of anti-fibrolysis genes in hepatocytes, endothelial cells, HSCs, and macrophages. Hepatocytes also had gene expression changes consistent with hepatocellular carcinoma. Collectively, these findings underscore the effects of TCDD in initiating SLD-like phenotypes and identified cell-specific gene expression changes related to oxidative stress, steatosis, fibrosis, cell proliferation and the development of HCC.
Collapse
Affiliation(s)
- Giovan N Cholico
- Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Rance Nault
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
- Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Tim Zacharewski
- Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA.
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA.
| |
Collapse
|
6
|
Ma R, Shi G, Li Y, Shi H. Trimethylamine N-oxide, choline and its metabolites are associated with the risk of non-alcoholic fatty liver disease. Br J Nutr 2024; 131:1915-1923. [PMID: 38443197 DOI: 10.1017/s0007114524000631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
It is inconclusive whether trimethylamine N-oxide (TMAO) and choline and related metabolites, namely trimethylamine (TMA), l-carnitine, betaine and dimethylglycine (DMG), are associated with non-alcoholic fatty liver disease (NAFLD). Our objective was to investigate these potential associations. Additionally, we sought to determine the mediating role of TMAO. In this 1:1 age- and sex-matched case-control study, a total of 150 pairs comprising NAFLD cases and healthy controls were identified. According to the fully adjusted model, after the highest tertile was compared with the lowest tertile, the plasma TMAO concentration (OR = 2·02 (95 % CI 1·04, 3·92); P trend = 0·003), l-carnitine concentration (OR = 1·79 (1·01, 3·17); P trend = 0·020) and DMG concentration (OR = 1·81 (1·00, 3·28); P trend = 0·014) were significantly positively associated with NAFLD incidence. However, a significantly negative association was found for plasma betaine (OR = 0. 50 (0·28, 0·88); P trend = 0·001). The restricted cubic splines model consistently indicated positive dose-response relationships between exposure to TMAO, l-carnitine, and DMG and NAFLD risk, with a negative association being observed for betaine. The corresponding AUC increased significantly from 0·685 (0·626, 0·745) in the traditional risk factor model to 0·769 (0·716, 0·822) when TMAO and its precursors were included (l-carnitine, betaine and choline) (P = 0·032). Mediation analyses revealed that 14·7 and 18·6 % of the excess NAFLD risk associated with l-carnitine and DMG, respectively, was mediated by TMAO (the P values for the mediating effects were 0·021 and 0·036, respectively). These results suggest that a higher concentration of TMAO is associated with increased NAFLD risk among Chinese adults and provide evidence of the possible mediating role of TMAO.
Collapse
Affiliation(s)
- Rong Ma
- Department of Infectious Diseases, the First Affiliated Hospital of Chengdu Medical College, Chengdu610500, People's Republic of China
| | - Guangying Shi
- Department of Hepatology, Xinjiang Corps Hospital, Xinjiang832104, People's Republic of China
| | - Yanfang Li
- Department of Infectious Diseases, the First Affiliated Hospital of Chengdu Medical College, Chengdu610500, People's Republic of China
| | - Han Shi
- Department of Infectious Diseases, the First Affiliated Hospital of Chengdu Medical College, Chengdu610500, People's Republic of China
| |
Collapse
|
7
|
Sugino KY, Janssen RC, McMahan RH, Zimmerman C, Friedman JE, Jonscher KR. Vertical Transfer of Maternal Gut Microbes to Offspring of Western Diet-Fed Dams Drives Reduced Levels of Tryptophan Metabolites and Postnatal Innate Immune Response. Nutrients 2024; 16:1808. [PMID: 38931163 PMCID: PMC11206590 DOI: 10.3390/nu16121808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Maternal obesity and/or Western diet (WD) is associated with an increased risk of metabolic dysfunction-associated steatotic liver disease (MASLD) in offspring, driven, in part, by the dysregulation of the early life microbiome. Here, using a mouse model of WD-induced maternal obesity, we demonstrate that exposure to a disordered microbiome from WD-fed dams suppressed circulating levels of endogenous ligands of the aryl hydrocarbon receptor (AHR; indole, indole-3-acetate) and TMAO (a product of AHR-mediated transcription), as well as hepatic expression of Il10 (an AHR target), in offspring at 3 weeks of age. This signature was recapitulated by fecal microbial transfer from WD-fed pregnant dams to chow-fed germ-free (GF) lactating dams following parturition and was associated with a reduced abundance of Lactobacillus in GF offspring. Further, the expression of Il10 was downregulated in liver myeloid cells and in LPS-stimulated bone marrow-derived macrophages (BMDM) in adult offspring, suggestive of a hypo-responsive, or tolerant, innate immune response. BMDMs from adult mice lacking AHR in macrophages exhibited a similar tolerogenic response, including diminished expression of Il10. Overall, our study shows that exposure to maternal WD alters microbial metabolites in the offspring that affect AHR signaling, potentially contributing to innate immune hypo-responsiveness and progression of MASLD, highlighting the impact of early life gut dysbiosis on offspring metabolism. Further investigations are warranted to elucidate the complex interplay between maternal diet, gut microbial function, and the development of neonatal innate immune tolerance and potential therapeutic interventions targeting these pathways.
Collapse
Affiliation(s)
- Kameron Y. Sugino
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
| | - Rachel C. Janssen
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
| | - Rachel H. McMahan
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Chelsea Zimmerman
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Karen R. Jonscher
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.Y.S.); (R.C.J.); (J.E.F.)
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
8
|
Agarwal M, Roth K, Yang Z, Sharma R, Maddipati K, Westrick J, Petriello MC. Loss of flavin-containing monooxygenase 3 modulates dioxin-like polychlorinated biphenyl 126-induced oxidative stress and hepatotoxicity. ENVIRONMENTAL RESEARCH 2024; 250:118492. [PMID: 38373550 PMCID: PMC11102846 DOI: 10.1016/j.envres.2024.118492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/21/2024]
Abstract
Dioxin-like pollutants (DLPs), such as polychlorinated biphenyl 126 (PCB 126), are synthetic chemicals classified as persistent organic pollutants. They accumulate in adipose tissue and have been linked to cardiometabolic disorders, including fatty liver disease. The toxicity of these compounds is associated with activation of the aryl hydrocarbon receptor (Ahr), leading to the induction of phase I metabolizing enzyme cytochrome P4501a1 (Cyp1a1) and the subsequent production of reactive oxygen species (ROS). Recent research has shown that DLPs can also induce the xenobiotic detoxification enzyme flavin-containing monooxygenase 3 (FMO3), which plays a role in metabolic homeostasis. We hypothesized whether genetic deletion of Fmo3 could protect mice, particularly in the liver, where Fmo3 is most inducible, against PCB 126 toxicity. To test this hypothesis, male C57BL/6 wild-type (WT) mice and Fmo3 knockout (Fmo3 KO) mice were exposed to PCB 126 or vehicle (safflower oil) during a 12-week study, at weeks 2 and 4. Various analyses were performed, including hepatic histology, RNA-sequencing, and quantitation of PCB 126 and F2-isoprostane concentrations. The results showed that PCB 126 exposure caused macro and microvesicular fat deposition in WT mice, but this macrovesicular fatty change was absent in Fmo3 KO mice. Moreover, at the pathway level, the hepatic oxidative stress response was significantly different between the two genotypes, with the induction of specific genes observed only in WT mice. Notably, the most abundant F2-isoprostane, 8-iso-15-keto PGE2, increased in WT mice in response to PCB 126 exposure. The study's findings also demonstrated that hepatic tissue concentrations of PCB 126 were higher in WT mice compared to Fmo3 KO mice. In summary, the absence of FMO3 in mice led to a distinctive response to dioxin-like pollutant exposure in the liver, likely due to alterations in lipid metabolism and storage, underscoring the complex interplay of genetic factors in the response to environmental toxins.
Collapse
Affiliation(s)
- Manisha Agarwal
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, 48202, USA; Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Katherine Roth
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Zhao Yang
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Rahul Sharma
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Krishnarao Maddipati
- Department of Pathology, Lipidomic Core Facility, Wayne State University, Detroit, MI, 48202, USA
| | - Judy Westrick
- Department of Chemistry, Lumigen Instrumentation Center, Wayne State University, Detroit, MI, 48202, USA
| | - Michael C Petriello
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, 48202, USA; Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA.
| |
Collapse
|
9
|
Huang Y, Zhang J, Zhang Y, Wang W, Li M, Chen B, Zhang X, Zhang Z, Huang J, Jin Y, Wang H, Zhang X, Yin S, Yang W. Red meat intake, faecal microbiome, serum trimethylamine N-oxide and hepatic steatosis among Chinese adults. Liver Int 2024; 44:1142-1153. [PMID: 38314906 DOI: 10.1111/liv.15860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND AND AIMS Emerging evidence suggests a detrimental impact of high red meat intake on hepatic steatosis. We investigated the potential interplay between red meat intake and gut microbiome on circulating levels of trimethylamine N-oxide (TMAO) and hepatic steatosis risk. METHODS This cross-sectional study was conducted in a representative sample of 754 community-dwelling adults in Huoshan, China. Diet was collected using 4 quarterly 3 consecutive 24-h dietary (12-day) recalls. We profiled faecal microbiome using 16S ribosomal RNA sequencing and quantified serum TMAO and its precursors using LC-tandem MS (n = 333). We detected hepatic steatosis by FibroScan. The adjusted odds ratios (aORs) and 95% confidence intervals (CIs) were calculated using logistic regression. RESULTS TMAO levels but not its precursors were positively associated with the likelihood of hepatic steatosis (aOR per 1-SD increment 1.86, 95% CI 1.04-3.32). We identified 14 bacterial genera whose abundance was associated with TMAO concentration (pFDR < .05) belonging to the phyla Firmicutes, Bacteroidetes, Actinobacteria and Proteobacteria families. Per 10 g/day increase in red meat intake was positively associated with TMAO levels among participants who had higher red meat intake (>70 g/day) and higher TMAO-predicting microbial scores (TMS, β = .045, p = .034), but not among others (pinteraction = .030). TMS significantly modified the positive association between red meat and steatosis (pinteraction = .032), with a stronger association being observed among participants with higher TMS (aOR 1.30, 95% CI 1.07-1.57). CONCLUSIONS The bacterial genera that predicted TMAO levels may jointly modify the association between red meat intake and TMAO levels and the subsequent risk of hepatic steatosis.
Collapse
Affiliation(s)
- Yong Huang
- School of Public Health, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Jiawei Zhang
- School of Public Health, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Yaozong Zhang
- School of Public Health, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Wuqi Wang
- School of Public Health, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Meiling Li
- School of Public Health, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Bo Chen
- School of Public Health, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Xiaoyu Zhang
- School of Public Health, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Zhuang Zhang
- School of Public Health, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Jiaqi Huang
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Yong Jin
- School of Public Health, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Hua Wang
- School of Public Health, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Xuehong Zhang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Shi Yin
- Department of Geriatrics, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Wanshui Yang
- School of Public Health, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| |
Collapse
|
10
|
Sheng M, Xu S, Chen WW, Li FQ, Zhong YM, Ouyang YX, Liao YL, Lai P. A bibliometric analysis of studies on the gut microbiota in cardiovascular disease from 2004 to 2022. Front Cell Infect Microbiol 2023; 12:1083995. [PMID: 36683688 PMCID: PMC9852829 DOI: 10.3389/fcimb.2022.1083995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/06/2022] [Indexed: 01/09/2023] Open
Abstract
Background Increasing evidence indicates that the gut microbiota (GM) is linked to cardiovascular disease (CVD). Many studies on the GM in CVD have been published in the last decade. However, bibliometric analysis in this field is still lacking. Methods On 30 September 2022, a search of the Web of Science™ (WoS; Clarivate™, Philadelphia, PA, USA) yielded 1,500 articles and reviews on the GM and CVD. Microsoft Excel and CiteSpace and VOSviewer software were used to analyze publication trends and research hotspots in this field. Results Our search generated 1,708 publications on the GM in CVD published between 2004 and 2022, and 1,500 articles and review papers were included in the final analysis. The number of publications relating to the GM in CVD increased from 1 in 2004 to 350 in 2021. China (485 publications, 9,728 non-self-citations, and an H-index of 47) and the USA (418 publications, 24,918 non-self-citations, and an H-index of 82) contributed 32.31%, and 27.85%, respectively, of the total number of publications. Examination of the number of publications (Np) and number of citations, excluding self-citations (Nc), of individual authors showed that Y. L. Tian (Np: 18, Nc: 262, and H-index: 12), from China, is the most productive author, followed by R. Knight (Np: 16, Nc: 3,036, and H-index: 15) and M. Nieuwdorp (Np: 16, Nc: 503, and H-index: 9). The Chinese Academy of Medical Sciences and Peking Union Medical College accounted for the largest number of publications (Np: 62, Nc: 3,727, and H-index: 13, average citation number (ACN): 60.11). The journal Nutrients had the most publications (Np: 73, Nc: 2,036, and ACN: 27.89). The emerging keywords in this field were "monooxygenase 3" (strength 3.24, 2020-2022), "short-chain fatty acid" (strength 4.63, 2021-2022), "fatty liver disease" (strength 3.18, 2021-2022), "metabolic disease" (strength 3.04, 2021-2022), "Mediterranean diet" (strength 2.95, 2021-2022), "prevention" (strength 2.77, 2021-2022), and "intestinal barrier" (strength 2.8, 2021-2022). Conclusion Publications on the GM in CVD rapidly increased in the last decade. The USA was the most influential country in publications in this field, followed by China. The journal with the most publications was Nutrients. Monooxygenase-3, short-chain fatty acids, fatty liver disease, metabolic disease, the Mediterranean diet, intestinal barrier, and prevention are the current hotspots or potential hotspots for future study.
Collapse
Affiliation(s)
- Ming Sheng
- Department of Library, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Shuquan Xu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Wei-Wei Chen
- Department of Pharmacology, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Fa-Quan Li
- Department of Cardiology, The First Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Yi-Ming Zhong
- Department of Cardiology, The First Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Yi-Xiang Ouyang
- Department of Library, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yong-Ling Liao
- Department of Cardiology, The First Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Ping Lai
- Department of Cardiology, The First Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| |
Collapse
|